Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cureus ; 12(1): e6547, 2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-32042522

RESUMEN

This case report describes a 57-year-old man who presented first with lethargy and dysarthria due to hyponatremia resulting from poor intake and diuretics. One week after discharge, he returned with confusion, ataxia and dysphagia, and he ultimately turned out to have developed an osmotic demyelination syndrome (ODS). In his first hospital admission, his serum sodium was corrected without new neurological symptoms occurring. In retrospect, he had several risk factors for the development of ODS during the correction of hyponatremia. The serum sodium correction rate only briefly exceeded the recommended limits. This case underlines that (1) extra awareness of the serum sodium correction rate is warranted in patients with risk factors, (2) factors other than sodium can play an important role in the development of ODS and (3) that the manifestations of ODS can be delayed substantially after an incident of osmotic stress.

2.
PLoS One ; 8(2): e55887, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23409077

RESUMEN

Non-invasive tubulointerstitial damage markers may allow better titration and monitoring of renoprotective therapy. We investigated the value of urinary vitamin D binding protein excretion (uVDBP) as a tubulointerstitial inflammation and fibrosis marker in adriamycin rats, and tested whether uVDBP parallels renal damage and responds to therapy intensification in humans. In adriamycin (ADR) rats, uVDBP was strongly elevated vs controls (CON) already 6 wks after nephrosis induction (ADR: 727±674 [mean±SD] vs CON: 9±12 µg/d, p<0.01), i.e. before onset of pre-fibrotic and inflammatory tubulointerstitial damage, and at all following 6-wk time points until end of follow up at 30 wks (ADR: 1403±1026 vs CON: 206±132 µg/d, p<0.01). In multivariate regression analysis, uVDBP was associated with tubulointerstitial macrophage accumulation (standardized beta = 0.47, p = 0.01) and collagen III expression (standardized beta = 0.44, p = 0.02) independently of albuminuria. In humans, uVDBP was increased in 100 microalbuminuric subjects (44±93 µg/d) and in 47 CKD patients with overt proteinuria (9.2±13.0 mg/d) compared to 100 normoalbuminuric subjects (12±12 µg/d, p<0.001). In CKD patients, uVDBP responded to intensification of renoprotective therapy (ACEi+liberal sodium: 9.2±13.0 mg/d vs dual RAAS blockade+low sodium: 2747±4013, p<0.001), but remained still >100-fold increased during maximal therapy vs normoalbuminurics (p<0.001), consistent with persisting tubulointerstitial damage. UVDBP was associated with tubular and inflammatory damage markers KIM-1 (standardized beta = 0.52, p<0.001), beta-2-microglobuline (st.beta = 0.45, p<0.001), cystatin C (st.beta = 0.40, p<0.001), MCP-1 (st.beta = 0.31, p<0.001) and NGAL (st.beta = 0.20, p = 0.005), independently of albuminuria. UVDBP may be a novel urinary biomarker of tubulointerstitial damage. Prospectively designed studies are required to validate our findings and confirm its relevance in the clinical setting.


Asunto(s)
Nefritis Intersticial/patología , Nefritis Intersticial/orina , Proteína de Unión a Vitamina D/orina , Albuminuria/orina , Animales , Biomarcadores/orina , Modelos Animales de Enfermedad , Doxorrubicina/efectos adversos , Fibrosis , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Nefritis Intersticial/inducido químicamente , Proteinuria/patología , Proteinuria/orina , Ratas
3.
PLoS One ; 7(11): e50209, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23189189

RESUMEN

Proteinuria is an important cause of progressive tubulo-interstitial damage. Whether proteinuria could trigger a renal lymphangiogenic response has not been established. Moreover, the temporal relationship between development of fibrosis, inflammation and lymphangiogenesis in chronic progressive kidney disease is not clear yet. Therefore, we evaluated the time course of lymph vessel (LV) formation in relation to proteinuria and interstitial damage in a rat model of chronic unilateral adriamycin nephrosis. Proteinuria and kidneys were evaluated up to 30 weeks after induction of nephrosis. LVs were identified by podoplanin/VEGFR3 double staining. After 6 weeks proteinuria was well-established, without influx of interstitial macrophages and myofibroblasts, collagen deposition, osteopontin expression (tubular activation) or LV formation. At 12 weeks, a ∼3-fold increase in cortical LV density was found (p<0.001), gradually increasing over time. This corresponded with a significant increase in tubular osteopontin expression (p<0.01) and interstitial myofibroblast numbers (p<0.05), whereas collagen deposition and macrophage numbers were not yet increased. VEGF-C was mostly expressed by tubular cells rather than interstitial cells. Cultured tubular cells stimulated with FCS showed a dose-dependent increase in mRNA and protein expression of VEGF-C which was not observed by human albumin stimulation. We conclude that chronic proteinuria provoked lymphangiogenesis in temporal conjunction with tubular osteopontin expression and influx of myofibroblasts, that preceded interstitial fibrosis.


Asunto(s)
Enfermedades Renales/etiología , Enfermedades Renales/patología , Linfangiogénesis , Proteinuria , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica , Humanos , Enfermedades Renales/genética , Túbulos Renales/patología , Lisinopril/administración & dosificación , Lisinopril/farmacología , Linfangiogénesis/efectos de los fármacos , Linfangiogénesis/genética , Vasos Linfáticos/patología , Masculino , Proteinuria/tratamiento farmacológico , Ratas , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo
4.
Ned Tijdschr Geneeskd ; 155(46): A3603, 2011.
Artículo en Holandés | MEDLINE | ID: mdl-22108460

RESUMEN

IgG4-related systemic disease is a new clinical entity with a large variety of clinical symptoms that can affect almost all organs. The best known manifestations are retroperitoneal fibrosis and autoimmune pancreatitis. We present 3 patients aged 71, 83 and 70 years, with malaise, fatigue and swellings suggestive of a malignancy. However, histopathology of these swellings showed infiltration with plasma cells. Increased serum IgG4-levels confirmed the diagnosis 'IgG4-related systemic disease'. All patients responded well to treatment with glucocorticoids. IgG4-related systemic disease is often mistaken for malignancy because of similar presenting symptoms. The diagnosis can easily be confirmed by high serum protein levels, high serum IgG4-levels and infiltrates of IgG4-positive plasma cells. Response to treatment with glucocorticoids is good, as is the prognosis. IgG4-related systemic disease should be part of the differential diagnosis when patients present with malaise, high protein-levels and multi-organ involvement. Rapid diagnosis can prevent unnecessary surgical procedures for malignancy.


Asunto(s)
Hipergammaglobulinemia/diagnóstico , Inmunoglobulina G/análisis , Células Plasmáticas/inmunología , Anciano , Anciano de 80 o más Años , Diagnóstico Diferencial , Glucocorticoides/uso terapéutico , Humanos , Hipergammaglobulinemia/complicaciones , Hipergammaglobulinemia/tratamiento farmacológico , Masculino , Neoplasias/diagnóstico , Células Plasmáticas/patología , Resultado del Tratamiento
5.
Nephrol Dial Transplant ; 24(9): 2637-45, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19429930

RESUMEN

BACKGROUND: Inhibition of the renin-angiotensin-aldosterone system (RAAS) provides renoprotection in adriamycin nephropathy (AN), along with a decrease in overexpression of glomerular heparanase. Angiotensin II (AngII) and reactive oxygen species (ROS) are known to regulate heparanase expression in vivo. However, it is unknown whether this is also the case for aldosterone. Therefore, we further assessed the role of aldosterone, AngII and ROS in the regulation of glomerular heparanase expression. METHODS: Six weeks after the induction of AN, rats were treated with vehicle (n = 8), lisinopril (75 mg/L, n = 10), spironolactone (3.3 mg/day, n = 12) or the combination of lisinopril and spironolactone (n = 14) for 12 weeks. Age-matched healthy rats served as controls (n = 6). After 18 weeks, renal heparanase and heparan sulfate (HS) expression were examined by immunofluorescence staining. In addition, the effect of aldosterone, AngII and ROS on heparanase expression in cultured podocytes was determined. RESULTS: Treatment with lisinopril, spironolactone or their combination significantly blunted the increased glomerular heparanase expression and restored the decreased HS expression in the GBM. Addition of aldosterone to cultured podocytes resulted in a significantly increased heparanase mRNA and protein expression, which could be inhibited by spironolactone. Heparanase mRNA and protein expression in podocytes were also significantly increased after stimulation with AngII or ROS. CONCLUSIONS: Our in vivo and in vitro results show that not only AngII and ROS, but also aldosterone is involved in the regulation of glomerular heparanase expression.


Asunto(s)
Aldosterona/farmacología , Angiotensina II/farmacología , Glucuronidasa/metabolismo , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/enzimología , Especies Reactivas de Oxígeno/farmacología , Aldosterona/metabolismo , Angiotensina II/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN/genética , Doxorrubicina/toxicidad , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucuronidasa/genética , Enfermedades Renales/inducido químicamente , Enfermedades Renales/enzimología , Enfermedades Renales/genética , Enfermedades Renales/patología , Glomérulos Renales/patología , Masculino , Podocitos/efectos de los fármacos , Podocitos/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Espironolactona/farmacología
6.
Am J Physiol Renal Physiol ; 296(5): F1136-45, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19225054

RESUMEN

Tubulointerstitial lesions are important in the progression of proteinuric renal disease. Tubular kidney injury molecule-1 (Kim-1) is induced in acute renal injury and reversible as a natural course. Kim-1 is also present in chronic renal damage; however, the dynamics of Kim-1 in chronic renal damage and effects of antiproteinuric treatment on Kim-1 are unknown. We studied Kim-1 in adriamycin nephrosis (AN) before and after renin-angiotensin system blockade. A renal biopsy was taken 6 wk after adriamycin injection to study renal damage and Kim-1 expression. Subsequently, ACE inhibition (ACEi; n = 23), angiotensin II antagonist (AT(1A); n = 23), or vehicle (n = 10) was given for 6 wk; healthy rats served as controls (CON; n = 8). In AN, renal Kim-1 mRNA was induced 26-fold vs. CON at week 6, with further increase in vehicle to week 12 (40-fold) but was reduced by ACEi and AT(1A) to 10- and 12-fold vs. CON (P < 0.05 vs. week 6). Kim-1 protein was undetectable in CON; in AN, it was present in brush border of dilated tubules in areas with adjacent interstitial lesions. Renal Kim-1 protein levels increased from weeks 6-12 in vehicle and decreased in ACEi- and AT(1A)-treated groups (P < 0.05). In vehicle, urinary Kim-1 was increased (P < 0.05 vs. CON), with a reduction by ACEi and AT(1A) (P < 0.05 vs. vehicle). Renal and urinary Kim-1 correlated with proteinuria and interstitial damage cross-sectionally. Reductions in proteinuria and renal Kim-1 correlated, which was not associated by corresponding changes in tubulointerstitial fibrosis. In conclusion, on longitudinal follow-up during antiproteinuric treatment increased renal Kim-1 expression is reversible in proportion to proteinuria reduction, likely reflecting reversibility of early tubular injury, supporting its potential as a biomarker for tubulointerstitial processes of damage and repair.


Asunto(s)
Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Túbulos Renales Proximales/fisiología , Proteinuria/fisiopatología , Insuficiencia Renal Crónica/fisiopatología , Animales , Antibióticos Antineoplásicos/toxicidad , Biomarcadores/metabolismo , Biopsia , Moléculas de Adhesión Celular/orina , Doxorrubicina/toxicidad , Fibrosis , Inmunohistoquímica , Túbulos Renales Proximales/patología , Masculino , Proteinuria/inducido químicamente , Proteinuria/patología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Insuficiencia Renal Crónica/inducido químicamente , Insuficiencia Renal Crónica/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Nephrol Dial Transplant ; 22(10): 2886-93, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17526541

RESUMEN

BACKGROUND: Minimal change nephrotic syndrome (MCNS) is the most frequent form of nephrotic syndrome in childhood. In the glomerular basement membrane (GBM) of adult patients with MCNS, a reduced expression of a specific heparan sulphate (HS) domain has been reported. In children with MCNS, urinary activity of the HS-degrading enzyme heparanase was increased. It is, therefore, possible that a decreased GBM HS expression is associated with the pathogenesis of proteinuria in patients with MCNS. METHODS: In this study, HS in glomeruli of five adult and six paediatric patients with MCNS were analysed by immunofluorescence staining using four different antibodies, each defining a specific sulphated HS domain. The pediatric patients were subdivided into three groups depending on the presence or absence of podocyte foot process effacement, the level of proteinuria and prednisone administration at the time of the biopsy. In addition, kidneys of rats with adriamycin nephropathy (ADRN), a model for MCNS, were included in the study. RESULTS: Expression of sulphated HS domains was not aberrant in adult or paediatric patients compared with control subjects. Children with and without proteinuria had the same HS content. In contrast, rats with ADRN showed a decreased glomerular expression of sulphated HS domains. CONCLUSIONS: These results suggest that in patients with MCNS proteinuria is not associated with major changes in glomerular expression of sulphated HS domains.


Asunto(s)
Regulación de la Expresión Génica , Glomérulos Renales/metabolismo , Síndrome Nefrótico/metabolismo , Adulto , Anciano , Animales , Biopsia , Niño , Preescolar , Doxorrubicina/farmacología , Femenino , Heparitina Sulfato/química , Humanos , Riñón/metabolismo , Riñón/patología , Masculino , Persona de Mediana Edad , Modelos Biológicos , Podocitos/metabolismo , Ratas , Ratas Wistar
9.
Am J Physiol Renal Physiol ; 292(1): F313-20, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16896183

RESUMEN

Kidney injury molecule-1 (Kim-1) is associated with ischemic and proteinuric tubular injury; however, whether dysregulation of the renin-angiotensin system (RAS) can also induce Kim-1 is unknown. We studied Kim-1 expression in homozygous Ren2 rats, characterized by renal damage through excessive RAS activation. We also investigated whether antifibrotic treatment (RAS blockade or p38 MAP kinase inhibition) would affect Kim-1 expression. At 7 wk of age, homozygous Ren2 rats received a nonhypotensive dose of candesartan (0.05 mg x kg(-1) x day(-1) sc) or the p38 inhibitor SB-239063 (15 mg x kg(-1) x day(-1) sc) for 4 wk; untreated Ren2 and Sprague-Dawley (SD) rats served as controls. Kim-1 mRNA and protein expression were determined by quantitative PCR and immunohistochemistry, respectively, and related to markers of prefibrotic renal damage. Urinary Kim-1 was measured in 8-wk-old Ren2 and SD rats with and without angiotensin-converting enzyme inhibition (ramipril, 1 mg x kg(-1) x day(-1) in drinking water for 4 wk). Untreated Ren2 rats showed a >20-fold increase in renal Kim-1 mRNA (expressed as Kim-1-to-GAPDH ratio): 75.5 +/- 43.6 vs. 3.1 +/- 1.0 in SD rats (P < 0.01). Candesartan and SB-239063 strongly reduced Kim-1 mRNA: 3.1 +/- 1.5 (P < 0.01) and 9.8 +/- 4.2 (P < 0.05), respectively. Kim-1 protein expression in damaged tubules paralleled mRNA expression. Kim-1 expression correlated with renal osteopontin, alpha-smooth muscle actin, and collagen III expression and with tubulointerstitial fibrosis. Damaged tubular segments expressing activated p38 also expressed Kim-1. Urinary Kim-1 was increased in Ren2 vs. SD (458 +/- 70 vs. 27 +/- 2 pg/ml, P < 0.01) rats and abolished in Ren2 rats treated with ramipril (33 +/- 5 pg/ml, P < 0.01). Kim-1 is associated with development of RAS-mediated renal damage. Antifibrotic treatment through RAS blockade or p38 MAP kinase inhibition reduced Kim-1 in the homozygous Ren2 model.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Proteínas de la Membrana/biosíntesis , Sistema Renina-Angiotensina/efectos de los fármacos , Renina/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Aldosterona/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Animales Modificados Genéticamente , Factor Natriurético Atrial/efectos de los fármacos , Factor Natriurético Atrial/fisiología , Bencimidazoles/farmacología , Biomarcadores , Compuestos de Bifenilo , Presión Sanguínea/fisiología , Creatinina/metabolismo , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Fibrosis , Inmunohistoquímica , Masculino , Nefritis Intersticial/metabolismo , Nefritis Intersticial/patología , Osteopontina/biosíntesis , Ratas , Ratas Endogámicas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tetrazoles/farmacología
10.
Nephron Physiol ; 103(1): p43-52, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16352916

RESUMEN

BACKGROUND: Antiproteinuric treatment by ACE inhibition (ACEi) provides renoprotection. However, resistance to antiproteinuric intervention occurs frequently, resulting in progressive renal damage. The extent of renal damage prior to treatment with ACEi reversely correlates with the antiproteinuric effects of ACEi in established adriamycin nephrosis. Sodium restriction enhances the response to ACEi, but whether it can overcome the negative predictive value of preceding renal damage on the therapeutic response is unknown. We studied the impact of preceding renal damage on the efficacy of ACEi in adriamycin nephrosis on different oral sodium loads. METHODS: Male Wistar rats were randomly assigned to a low (LS), normal (NS) or high (HS) sodium diet, initiated 1 week before adriamycin induction. At week 6, proteinuria was stabilized (195 +/- 172 mg/24 h), a renal biopsy was performed for analysis of preceding damage and rats were instituted on lisinopril for 6 weeks until sacrifice at week 12. RESULTS: ACEi reduced proteinuria in LS and NS animals. On univariate analysis, the antiproteinuric response was significantly predicted by preceding renal damage (focal glomerulosclerosis, interstitial macrophages and interstitial alpha-smooth muscle cell actin expression). On multivariate analysis, both sodium intake and preceding renal damage independently predicted residual proteinuria during ACEi (R2 model: 80% and 75% for data after 3 and 6 weeks of therapy, respectively). CONCLUSION: Our data confirm the predictive value of pretreatment renal damage for the antiproteinuric response to ACEi, despite the fact that the renal damage prior to the ACEi was very mild. The impact of pretreatment damage on the therapeutic response, however, was overcome by low sodium. Thus, the impact of pretreatment damage does not warrant therapeutic nihilism, but rather optimization of therapy response by dietary sodium restriction. Further studies are needed to elucidate whether this also applies to more severe damage, and whether combining ACEi with low sodium diet can improve long-term renal outcome in human.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Doxorrubicina/toxicidad , Enfermedades Renales/fisiopatología , Proteinuria/prevención & control , Sodio en la Dieta/uso terapéutico , Animales , Enfermedades Renales/inducido químicamente , Masculino , Pronóstico , Proteinuria/inducido químicamente , Proteinuria/diagnóstico , Ratas , Ratas Wistar
11.
J Pathol ; 207(4): 483-92, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16211543

RESUMEN

Proteinuria is associated with macrophage-dependent interstitial fibrosis (IF). Osteopontin (OPN), a macrophage chemoattractant, may be involved in the transition of proteinuria to IF but protective properties have also been reported. To elucidate whether OPN may be involved in the proteinuria-induced cascade of tubulointerstitial damage, renal expression of OPN was studied during the development of proteinuria-induced renal damage and during anti-proteinuric intervention with ACE inhibition (ACEi). First, the temporal relationships between proteinuria, interstitial OPN induction, and IF in adriamycin nephrosis (AN), a model of chronic proteinuria-induced renal damage, were studied. Second, the effect of anti-proteinuric treatment on OPN expression was investigated. The time course of OPN induction and markers of renal damage was studied in rats with unilateral AN at 6-week intervals until week 30. In a second study, a renal biopsy was taken 6 weeks after induction of bilateral AN; subsequently, rats were treated with ACEi until termination (week 12). In unilateral AN, proteinuria developed gradually and stabilized at week 10. In proteinuric kidneys, OPN expression was induced from week 12 onwards. Simultaneously, a progressive increase in interstitial macrophages, alpha-smooth muscle actin (alpha-SMA), collagen type III, and focal glomerulosclerosis (FGS) was observed. In bilateral AN, ACEi reduced proteinuria and OPN protein and stabilized fibrosis. In untreated animals, OPN mRNA increased, with stable OPN protein and fibrosis and increased FGS. Thus, in AN, development of proteinuria is followed by up-regulation of OPN along with markers of renal damage. The up-regulation of OPN is reversible by anti-proteinuric treatment without a corresponding reduction in fibrosis. Whereas these data are consistent with a role for OPN in the cascade of transition from proteinuria to fibrosis, intervention with ACEi showed that reduction of OPN does not attenuate established fibrosis.


Asunto(s)
Nefritis Intersticial/metabolismo , Proteinuria/metabolismo , Sialoglicoproteínas/fisiología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Colágeno Tipo III/metabolismo , Progresión de la Enfermedad , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Hibridación in Situ , Túbulos Renales/patología , Macrófagos/patología , Masculino , Nefritis Intersticial/tratamiento farmacológico , Nefritis Intersticial/patología , Osteopontina , Proteinuria/tratamiento farmacológico , ARN Mensajero/genética , Ratas , Ratas Wistar , Sialoglicoproteínas/biosíntesis , Sialoglicoproteínas/genética , Regulación hacia Arriba
12.
Nephrol Dial Transplant ; 20(10): 2060-70, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15956058

RESUMEN

BACKGROUND: Advanced glycation end-products (AGEs) contribute to the pathogenesis of diabetic glomerulopathy. The role of AGEs in non-diabetic renal damage is not well characterized. First, we studied whether renal AGE accumulation occurs in non-diabetic proteinuria-induced renal damage and whether this is ameliorated by renoprotective treatment. Secondly, we investigated whether renal AGE accumulation was due to intrarenal effects of local protein trafficking. METHODS: Pentosidine was measured (by high-performance liquid chromatography) in rats with chronic bilateral adriamycin nephropathy (AN), untreated and treated with lisinopril. Age-matched healthy rats served as negative controls. Secondly, we compared renal pentosidine in mild proteinuric and non-proteinuric kidneys of unilateral AN and in age-matched controls at 12 and 30 weeks. Intrarenal localization of pentosidine was studied by immunohistochemistry. RESULTS: Renal pentosidine was elevated in untreated AN (0.14+/-0.04 micromol/mol valine) vs healthy controls (0.04+/-0.01 micromol/mol valine, P<0.01). In lisinopril-treated AN, pentosidine was lower (0.09+/-0.02 micromol/mol valine) than in untreated AN (P<0.05). In unilateral proteinuria, pentosidine was similar in non-proteinuric and proteinuric kidneys. After 30 weeks of unilateral proteinuria, pentosidine was increased in both kidneys (0.26+/-0.10 micromol/mol valine) compared with controls (0.18+/-0.06 micromol/mol valine, P<0.05). Pentosidine (AN, week 30) was also increased compared with AN at week 12 (0.16+/-0.06 micromol/mol valine, P<0.01). In control and diseased kidneys, pentosidine was present in the collecting ducts. In proteinuric kidneys, in addition, pentosidine was present in the brush border and cytoplasm of dilated tubular structures, i.e. at sites of proteinuria-induced tubular damage. CONCLUSION: Pentosidine accumulates in non-diabetic proteinuric kidneys in damaged tubules, and renoprotective treatment by angiotensin-converting enzyme (ACE) inhibitors inhibits AGE accumulation, supporting a relationship between abnormal renal protein trafficking, proteinuria-induced tubular damage and tubular pentosidine accumulation. Future studies, applying specific AGE inhibitors, should be conducted to provide insight into the pathophysiological significance of renal AGEs in non-diabetic renal disease.


Asunto(s)
Arginina/análogos & derivados , Productos Finales de Glicación Avanzada/metabolismo , Riñón/metabolismo , Lisina/análogos & derivados , Proteinuria/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Arginina/metabolismo , Presión Sanguínea , Colesterol/sangre , Creatinina/orina , Doxorrubicina/toxicidad , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/patología , Lisinopril/farmacología , Lisina/metabolismo , Masculino , Proteinuria/inducido químicamente , Proteinuria/tratamiento farmacológico , Proteinuria/patología , Ratas , Ratas Wistar
13.
Nephrol Dial Transplant ; 20(1): 59-64, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15572383

RESUMEN

BACKGROUND: In man, differences in angiotensin-converting enzyme (ACE) levels, related to ACE (I/D) genotype, are associated with renal prognosis. This raises the hypothesis that individual differences in renal ACE activity are involved in renal susceptibility to inflicted damage. Therefore, we studied the predictive effect of renal ACE activity for the severity of renal damage induced by a single injection of adriamycin in rats. METHODS: Renal ACE activity (Hip-His-Leu cleavage by cortical homogenates) was determined by renal biopsy in 27 adult male Wistar rats. After 1 week of recovery, proteinuria was induced by adriamycin [1.5 mg/kg intravenously (i.v.) n = 18; controls, saline i.v. n = 9]. Proteinuria was measured every 2 weeks. After 12 weeks, rats were sacrificed and their kidneys harvested. RESULTS: As anticipated, adriamycin elicited nephrotic range proteinuria, renal interstitial damage and mild focal glomerulosclerosis. Baseline renal ACE positively correlated with the relative rise in proteinuria after adriamycin (r = 0.62, P<0.01), renal interstitial alpha-smooth muscle actin (r = 0.49, P<0.05), interstitial macrophage influx (r = 0.56, P<0.05), interstitial collagen III (r = 0.53, P<0.05), glomerular alpha-smooth muscle actin (r = 0.74, P<0.01) and glomerular desmin (r = 0.48, P<0.05). Baseline renal ACE did not correlate with focal glomerulosclerosis (r = 0.22, NS). In controls, no predictive values for renal parameters were observed. CONCLUSION: Individual differences in renal ACE activity predict the severity of adriamycin-induced renal damage in this outbred rat strain. This supports the assumption that differences in renal ACE activity predispose to a less favourable course of renal damage.


Asunto(s)
Doxorrubicina/toxicidad , Riñón/efectos de los fármacos , Riñón/patología , Nefrosis/patología , Peptidil-Dipeptidasa A/metabolismo , Animales , Biopsia con Aguja , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Inmunohistoquímica , Masculino , Nefrosis/inducido químicamente , Peptidil-Dipeptidasa A/análisis , Valor Predictivo de las Pruebas , Ratas , Ratas Wistar , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas
14.
Thyroid ; 14(7): 544-7, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15307945

RESUMEN

Subacute thyroiditis (SAT) is a spontaneously remitting inflammatory disorder of the thyroid, associated with human leukocyte antigen (HLA)-B35, and may be virally induced in genetically predisposed individuals. A 57-year-old Caucasian man presented with symptoms of hyperthyroidism as well as enlargement and tenderness of the thyroid. The patient had an elevated erythrocyte sedimentation rate, low thyrotropin (TSH) and elevated thyroxine and triiodothyroinine levels with suppressed 131I thyroidal uptake. He was diagnosed to have SAT. In the patient's family three sisters and one brother also had had SAT, as probably did the deceased father. Because of the familial occurrence HLA-typing was performed. All affected family members were heterozygous for HLA-B35. The family members lived more than 50 miles apart in different regions of The Netherlands and had SAT at different time points between 1986 and 2002, which in combination with HLA-B35 seems to highlight the importance of genetic influences as a risk factor for the development of SAT in this family. In conclusion, the case described here represents the second familial incidence and largest family reported so far with occurrence of SAT in association with HLA-B35.


Asunto(s)
Antígeno HLA-B35/genética , Tiroiditis Subaguda/genética , Anciano , Anciano de 80 o más Años , Salud de la Familia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje
15.
J Pathol ; 201(1): 160-7, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12950029

RESUMEN

ACE inhibition (ACEi) reduces proteinuria and provides reno-protection, but not all subjects benefit from ACEi. Individual differences in the reduction in proteinuria at the onset of treatment and in residual proteinuria during therapy predict differences in renal outcome. The present study investigated whether individual differences in the anti-proteinuric efficacy of ACEi are explained by differences in the severity of pretreatment renal structural damage and whether differences in the level of residual proteinuria during therapy are explained by the severity of renal structural damage at that time, in adriamycin nephrosis in the rat. Pretreatment renal structural damage was assessed in biopsies 6 weeks after exposure to adriamycin (2 mg/kg iv). Then ACEi (75 mg/l lisinopril, n = 23) or vehicle (n = 10) was administered; renal biopsies were repeated after stabilization of the anti-proteinuric response (week 8). Early renal damage (interstitial alpha-smooth muscle actin expression and macrophage accumulation) and established lesions [focal glomerulosclerosis (FGS) and interstitial fibrosis] were scored. During ACEi, proteinuria fell from 834 (487-851) mg/24 h pretreatment to 153 (66-265) mg/24 h at week 8 (p < 0.05); FGS stabilized from 27 (4-70) arbitrar units (AU) pretreatment to 26 (4-84) at week 12, whereas the vehicle did not affect proteinuria, resulting in progressive FGS: 18 (10-26) AU pretreatment versus 88 (46-130) at week 12 (p < 0.05). All parameters of pretreatment damage significantly predicted the anti-proteinuric response. Residual proteinuria during ACEi correlated significantly with renal structural damage parameters at that time. Pretreatment renal damage also predicted renal outcome during extended treatment. Thus, in this experimental setting, in rats with the same renal disorder and the same duration of disease, individual differences in pretreatment renal damage, albeit relatively modest, explain individual differences in renal responsiveness to ACEi. This implies that the limits of the efficacy of ACEi are set by prevalent renal damage. Further studies into the mechanisms of individual resistance to the anti-proteinuric action of ACEi are needed to develop additive intervention strategies.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Nefrosis/tratamiento farmacológico , Proteinuria/tratamiento farmacológico , Actinas/metabolismo , Animales , Antibióticos Antineoplásicos , Doxorrubicina , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/patología , Macrófagos/patología , Masculino , Nefrosis/inducido químicamente , Nefrosis/patología , Pronóstico , Proteinuria/inducido químicamente , Proteinuria/patología , Ratas , Ratas Wistar , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...